101
|
Hori K, Ajioka K, Goda N, Shindo A, Takagishi M, Tenno T, Hiroaki H. Discovery of Potent Disheveled/Dvl Inhibitors Using Virtual Screening Optimized With NMR-Based Docking Performance Index. Front Pharmacol 2018; 9:983. [PMID: 30233369 PMCID: PMC6134994 DOI: 10.3389/fphar.2018.00983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 08/10/2018] [Indexed: 12/13/2022] Open
Abstract
Most solid tumors have their own cancer stem cells (CSCs), which are resistant to standard chemo-therapies. Recent reports have described that Wnt pathway plays a key role in self-renewal and tumorigenesis of CSCs. Regarding the Wnt/β-catenin pathway, Dvl (mammalian Disheveled) is an attractive target of drug discovery. After analyzing the PDZ domain of human Dvl1 (Dvl1-PDZ) using NMR, we subjected it to preliminary NMR titration studies with 17 potential PDZ-binding molecules including CalBioChem-322338, a commercially available Dvl PDZ domain inhibitor. Next, we performed virtual screening (VS) using the program GOLD with nine parameter sets. Results were evaluated using the NMR-derived docking performance index (NMR-DPI). One parameter set of GOLD docking showing the best NMR-DPI was selected and used for the second VS against 5,135 compounds. The second docking trial identified more than 1,700 compounds that exhibited higher scores than CalBioChem-322338. Subsequent NMR titration experiments with five new candidate molecules (NPL-4001, 4004, 4011, 4012, and 4013), Dvl1-PDZ revealed larger chemical shift changes than those of CalBioChem-322338. Finally, these compounds showed partial proliferation inhibition activity against BT-20, a triple negative breast cancer (TNBC) cell. These compounds are promising Wnt pathway inhibitors that are potentially useful for anti-TNBC therapy.
Collapse
Affiliation(s)
- Kiminori Hori
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Kasumi Ajioka
- Department of Biological Science, School of Science, Nagoya University, Nagoya, Japan
| | - Natsuko Goda
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Asako Shindo
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Maki Takagishi
- Department of Pathology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Takeshi Tenno
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan.,BeCellBar LLC, Business Incubation Center, Nagoya University, Nagoya, Japan
| | - Hidekazu Hiroaki
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan.,Department of Biological Science, School of Science, Nagoya University, Nagoya, Japan.,BeCellBar LLC, Business Incubation Center, Nagoya University, Nagoya, Japan
| |
Collapse
|
102
|
Quan H, Li B, Yang J. MicroRNA-504 functions as a tumor suppressor in hepatocellular carcinoma through inhibiting Frizzled-7-mediated-Wnt/β-catenin signaling. Biomed Pharmacother 2018; 107:754-762. [PMID: 30142536 DOI: 10.1016/j.biopha.2018.07.150] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/11/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests that microRNAs (miRNAs) are critical regulators in the development and progression of various malignant tumors, including hepatocellular carcinoma (HCC). Multiple findings have indicated that miRNA-504 (miR-504) is dysregulated in several types of cancers, functioning as an oncogenic miRNA or a tumor suppressive miRNA. However, the role of miR-504 in HCC remains unknown. In this study, we aimed to detect the expression pattern of miR-504 in HCC tissues and cell lines and investigate the precise biological function in HCC cells. Our results showed that miR-504 expression levels were frequently downregulated in both HCC tissues and cell lines. Gain-of-function experiments demonstrated that miR-504 overexpression inhibited the proliferation and invasion in HCC cell lines. By contrast, miR-504 inhibition had the opposite effect. Interestingly, bioinformatics analysis predicted that Frizzled-7 (FZD7) was a potential target gene of miR-504. Dual-luciferase reporter assays confirmed that miR-504 directly targeted the 3'-untranslated region of FZD7 mRNA. In addition, our results showed that miR-504 negatively regulated the mRNA and protein expression of FZD7 in HCC cell lines. Moreover, miR-540 overexpression inhibited the cellular expression of β-catenin and blocked the activation of Wnt signaling in HCC cells. Notably, restoration of FZD7 expression significantly reversed the inhibitory effect of miR-504 on proliferation, invasion, and Wnt/β-catenin signaling in HCC cells. In conclusion, our results demonstrate that miR-504 functions as a tumor suppressive miRNA that inhibits the proliferation and invasion of HCC cells by targeting FZD7 and inhibiting Wnt/β-catenin signaling. Our study provides evidence that miR-504-meidated FZD7/Wnt/β-catenin signaling pathway plays an important role in HCC development and progression and suggests miR-504 as a novel future therapeutic target for treatment of HCC.
Collapse
Affiliation(s)
- Hui Quan
- Department of Interventional Vascular Surgery, Baoji Central Hospital, Baoji, Shaanxi Province 721008, China
| | - Bo Li
- Department of Interventional Radiology, Traditional Chinese Medical Hospital of Baoji City, Baoji, Shaanxi Province 721001, China
| | - Jianjun Yang
- Department of Infectious Diseases, Baoji Central Hospital, No. 8 Jiangtan Road, Weibin District, Baoji, Shaanxi Province 721008, China.
| |
Collapse
|
103
|
Koval A, Pieme CA, Queiroz EF, Ragusa S, Ahmed K, Blagodatski A, Wolfender JL, Petrova TV, Katanaev VL. Tannins from Syzygium guineense suppress Wnt signaling and proliferation of Wnt-dependent tumors through a direct effect on secreted Wnts. Cancer Lett 2018; 435:110-120. [PMID: 30098400 DOI: 10.1016/j.canlet.2018.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 06/12/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancer (TNBC) and colon cancer (CC) are two stigmatic examples of poorly treatable tumors, whose progression critically depends upon hyperactivation of the Wnt signaling. Development of specific anti-Wnt inhibitors is required to develop drugs against these and other Wnt-dependent cancers. Natural products, especially plants, have been used for the treatment of various diseases from ancient times. We examined extracts from several indigenous Cameroonian herbs and tested their effects on proliferation and Wnt signaling in TNBC and CC cells. Extracts from "fruit rouge", Syzygium guineense Wall. (Myrtaceae), demonstrated a strong activity against these cancer cells, as well as CC organoids. We found TNBC cells to significantly upregulate expression of Wnt3a, and the effects of S. guineense extracts on TNBC cell proliferation correlated with inhibition of the Wnt3a-induced β-catenin stabilization and transcriptional response. HPLC analysis revealed that the active components belong to tannins. We found a direct destabilizing effect of S. guineense extract on Wnt3a and other Wnt proteins, identifying a novel mechanism of action of tannins on the Wnt signaling pathway and cancer cell proliferation. Being edible, this African plant may have an important cancer-preventive nutritional value.
Collapse
Affiliation(s)
- Alexey Koval
- Department of Pharmacology and Toxicology, University of Lausanne, Switzerland
| | - Constant A Pieme
- Department of Pharmacology and Toxicology, University of Lausanne, Switzerland; Department of Biochemistry and Physiological Sciences, University of Yaounde I, Cameroon
| | | | - Simone Ragusa
- (d)Department of Oncology, Ludwig Cancer Research, Lausanne Branch, and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Kamal Ahmed
- Department of Pharmacology and Toxicology, University of Lausanne, Switzerland
| | - Artem Blagodatski
- Department of Pharmacology and Toxicology, University of Lausanne, Switzerland; School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | | | - Tatiana V Petrova
- (d)Department of Oncology, Ludwig Cancer Research, Lausanne Branch, and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Vladimir L Katanaev
- Department of Pharmacology and Toxicology, University of Lausanne, Switzerland; School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia.
| |
Collapse
|
104
|
A Comparative Study on Anti-Invasion, Antimigration, and Antiadhesion Effects of the Bioactive Carotenoids of Saffron on 4T1 Breast Cancer Cells Through Their Effects on Wnt/β-Catenin Pathway Genes. DNA Cell Biol 2018; 37:697-707. [DOI: 10.1089/dna.2018.4248] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
105
|
Dittmer J. Breast cancer stem cells: Features, key drivers and treatment options. Semin Cancer Biol 2018; 53:59-74. [PMID: 30059727 DOI: 10.1016/j.semcancer.2018.07.007] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
The current view is that breast cancer is a stem cell disease characterized by the existence of cancer cells with stem-like features and tumor-initiating potential. These cells are made responsible for tumor dissemination and metastasis. Common therapies by chemotherapeutic drugs fail to eradicate these cells and rather increase the pool of cancer stem cells in tumors, an effect that may increase the likelyhood of recurrence. Fifteen years after the first evidence for a small stem-like subpopulation playing a major role in breast cancer initiation has been published a large body of knowledge has been accumulated regarding the signaling cascades and proteins involved in maintaining stemness in breast cancer. Differences in the stem cell pool size and in mechanisms regulating stemness in the different breast cancer subtypes have emerged. Overall, this knowledge offers new approaches to intervene with breast cancer stem cell activity. New options are particularly needed for the treatment of triple-negative breast cancer subtype, which is particularly rich in cancer stem cells and is also the subtype for which specific therapies are still not available.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
106
|
Ji J, He D, Feng Y, He Y, Xue F, Xie L. JDINAC: joint density-based non-parametric differential interaction network analysis and classification using high-dimensional sparse omics data. Bioinformatics 2018; 33:3080-3087. [PMID: 28582486 DOI: 10.1093/bioinformatics/btx360] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 06/01/2017] [Indexed: 12/26/2022] Open
Abstract
Motivation A complex disease is usually driven by a number of genes interwoven into networks, rather than a single gene product. Network comparison or differential network analysis has become an important means of revealing the underlying mechanism of pathogenesis and identifying clinical biomarkers for disease classification. Most studies, however, are limited to network correlations that mainly capture the linear relationship among genes, or rely on the assumption of a parametric probability distribution of gene measurements. They are restrictive in real application. Results We propose a new Joint density based non-parametric Differential Interaction Network Analysis and Classification (JDINAC) method to identify differential interaction patterns of network activation between two groups. At the same time, JDINAC uses the network biomarkers to build a classification model. The novelty of JDINAC lies in its potential to capture non-linear relations between molecular interactions using high-dimensional sparse data as well as to adjust confounding factors, without the need of the assumption of a parametric probability distribution of gene measurements. Simulation studies demonstrate that JDINAC provides more accurate differential network estimation and lower classification error than that achieved by other state-of-the-art methods. We apply JDINAC to a Breast Invasive Carcinoma dataset, which includes 114 patients who have both tumor and matched normal samples. The hub genes and differential interaction patterns identified were consistent with existing experimental studies. Furthermore, JDINAC discriminated the tumor and normal sample with high accuracy by virtue of the identified biomarkers. JDINAC provides a general framework for feature selection and classification using high-dimensional sparse omics data. Availability and implementation R scripts available at https://github.com/jijiadong/JDINAC. Contact lxie@iscb.org. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jiadong Ji
- Department of Mathematical Statistics, School of Statistics, Shandong University of Finance and Economics, Jinan 250014, China
| | - Di He
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Yang Feng
- Department of Statistics, Columbia University, New York, NY 10027, USA
| | - Yong He
- Department of Mathematical Statistics, School of Statistics, Shandong University of Finance and Economics, Jinan 250014, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, Jinan 250012, China
| | - Lei Xie
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY 10016, USA.,Department of Computer Science, Hunter College, The City University of New York, NY 10065, USA
| |
Collapse
|
107
|
Frizzled Receptors as Potential Therapeutic Targets in Human Cancers. Int J Mol Sci 2018; 19:ijms19051543. [PMID: 29789460 PMCID: PMC5983605 DOI: 10.3390/ijms19051543] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/12/2018] [Accepted: 05/19/2018] [Indexed: 12/14/2022] Open
Abstract
Frizzled receptors (FZDs) are a family of seven-span transmembrane receptors with hallmarks of G protein-coupled receptors (GPCRs) that serve as receptors for secreted Wingless-type (WNT) ligands in the WNT signaling pathway. Functionally, FZDs play crucial roles in regulating cell polarity, embryonic development, cell proliferation, formation of neural synapses, and many other processes in developing and adult organisms. In this review, we will introduce the basic structural features and review the biological function and mechanism of FZDs in the progression of human cancers, followed by an analysis of clinical relevance and therapeutic potential of FZDs. We will focus on the development of antibody-based and small molecule inhibitor-based therapeutic strategies by targeting FZDs for human cancers.
Collapse
|
108
|
Preferential Inhibition of Wnt/β-Catenin Signaling by Novel Benzimidazole Compounds in Triple-Negative Breast Cancer. Int J Mol Sci 2018; 19:ijms19051524. [PMID: 29783777 PMCID: PMC5983770 DOI: 10.3390/ijms19051524] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/09/2018] [Accepted: 05/15/2018] [Indexed: 12/31/2022] Open
Abstract
Wnt/β-catenin signaling is upregulated in triple-negative breast cancer (TNBC) compared to other breast cancer subtypes and normal tissues. Current Wnt/β-catenin inhibitors, such as niclosamide, target the pathway nonspecifically and exhibit poor pharmacokinetics/pharmacodynamics in vivo. Niclosamide targets other pathways, including mTOR, STAT3 and Notch. Novel benzimidazoles have been developed to inhibit Wnt/β-catenin signaling with greater specificity. The compounds SRI33576 and SRI35889 were discovered to produce more cytotoxicity in TNBC cell lines than in noncancerous cells. The agents also downregulated Wnt/β-catenin signaling mediators LRP6, cyclin D1, survivin and nuclear active β-catenin. In addition, SRI33576 did not affect mTOR, STAT3 and Notch signaling in TNBC and noncancerous cells. SRI35889 inhibited mTOR signaling less in noncancerous than in cancerous cells, while not affecting STAT3 and Notch pathways. Compounds SRI32529, SRI35357 and SRI35361 were not selectively cytotoxic against TNBC cell lines compared to MCF10A cells. While SRI32529 inhibited Wnt/β-catenin signaling, the compound also mitigated mTOR, STAT3 and Notch signaling. SRI33576 and SRI35889 were identified as cytotoxic and selective inhibitors of Wnt/β-catenin signaling with therapeutic potential to treat TNBC in vivo.
Collapse
|
109
|
Abstract
Wnt signaling is important for breast development and remodeling during pregnancy and lactation. Epigenetic modifications change expression levels of components of the Wnt pathway, underlying oncogenic transformation. However, no clear Wnt component increasing expression universally across breast cancer (BC) or its most Wnt-dependent triple-negative BC (TNBC) subgroup has been identified, delaying development of targeted therapies. Here we perform network correlation analysis of expression of >100 Wnt pathway components in hundreds of healthy and cancerous breast tissues. Varying in expression levels among people, Wnt components remarkably coordinate their production; this coordination is dramatically decreased in BC. Clusters with coordinated gene expression exist within the healthy cohort, highlighting Wnt signaling subtypes. Different BC subgroups are identified, characterized by different remaining Wnt signaling signatures, providing the rational for patient stratification for personalizing the therapeutic applications. Key pairwise interactions within the Wnt pathway (some inherited and some established de novo) emerge as targets for future drug discovery against BC.
Collapse
|
110
|
Maubant S, Tahtouh T, Brisson A, Maire V, Némati F, Tesson B, Ye M, Rigaill G, Noizet M, Dumont A, Gentien D, Marty-Prouvost B, de Koning L, Mahmood SF, Decaudin D, Cruzalegui F, Tucker GC, Roman-Roman S, Dubois T. LRP5 regulates the expression of STK40, a new potential target in triple-negative breast cancers. Oncotarget 2018; 9:22586-22604. [PMID: 29854300 PMCID: PMC5978250 DOI: 10.18632/oncotarget.25187] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancers (TNBCs) account for a large proportion of breast cancer deaths, due to the high rate of recurrence from residual, resistant tumor cells. New treatments are needed, to bypass chemoresistance and improve survival. The WNT pathway, which is activated in TNBCs, has been identified as an attractive pathway for treatment targeting. We analyzed expression of the WNT coreceptors LRP5 and LRP6 in human breast cancer samples. As previously described, LRP6 was overexpressed in TNBCs. However, we also showed, for the first time, that LRP5 was overexpressed in TNBCs too. The knockdown of LRP5 or LRP6 decreased tumorigenesis in vitro and in vivo, identifying both receptors as potential treatment targets in TNBC. The apoptotic effect of LRP5 knockdown was more robust than that of LRP6 depletion. We analyzed and compared the transcriptomes of cells depleted of LRP5 or LRP6, to identify genes specifically deregulated by LRP5 potentially implicated in cell death. We identified serine/threonine kinase 40 (STK40) as one of two genes specifically downregulated soon after LRP5 depletion. STK40 was found to be overexpressed in TNBCs, relative to other breast cancer subtypes, and in various other tumor types. STK40 depletion decreased cell viability and colony formation, and induced the apoptosis of TNBC cells. In addition, STK40 knockdown impaired growth in an anchorage-independent manner in vitro and slowed tumor growth in vivo. These findings identify the largely uncharacterized putative protein kinase STK40 as a novel candidate treatment target for TNBC.
Collapse
Affiliation(s)
- Sylvie Maubant
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Tania Tahtouh
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Amélie Brisson
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Virginie Maire
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Fariba Némati
- Institut Curie, PSL Research University, Translational Research Department, Preclinical Investigation Laboratory, Paris, France
| | - Bruno Tesson
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France.,Institut Curie, PSL Research University, INSERM U900, Paris, France
| | - Mengliang Ye
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Guillem Rigaill
- Institute of Plant Sciences Paris-Saclay (IPS2), UMR 9213/UMR 1403, CNRS, INRA, Université Paris-Sud, Université d'Evry, Université Paris-Diderot, Sorbonne Paris-Cité, Orsay, France.,Laboratoire de Mathématiques et Modélisation d'Evry (LaMME), Université d'Evry Val d'Essonne, UMR CNRS 8071, ENSIIE, USC INRA, Évry, France
| | - Maïté Noizet
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Aurélie Dumont
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - David Gentien
- Institut Curie, PSL Research University, Translational Research Department, Genomics Platform, Paris, France
| | - Bérengère Marty-Prouvost
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Leanne de Koning
- Institut Curie, PSL Research University, Translational Research Department, Reverse-Phase Protein Array Platform, Paris, France
| | - Sardar Faisal Mahmood
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| | - Didier Decaudin
- Institut Curie, PSL Research University, Translational Research Department, Preclinical Investigation Laboratory, Paris, France
| | - Francisco Cruzalegui
- Oncology Research and Development Unit, Institut de Recherches SERVIER, Croissy-Sur-Seine, France
| | - Gordon C Tucker
- Oncology Research and Development Unit, Institut de Recherches SERVIER, Croissy-Sur-Seine, France
| | - Sergio Roman-Roman
- Institut Curie, PSL Research University, Translational Research Department, Paris, France
| | - Thierry Dubois
- Institut Curie, PSL Research University, Translational Research Department, Breast Cancer Biology Group, Paris, France
| |
Collapse
|
111
|
Chen Z, Huang C, Ma T, Jiang L, Tang L, Shi T, Zhang S, Zhang L, Zhu P, Li J, Shen A. Reversal effect of quercetin on multidrug resistance via FZD7/β-catenin pathway in hepatocellular carcinoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 43:37-45. [PMID: 29747752 DOI: 10.1016/j.phymed.2018.03.040] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/26/2018] [Accepted: 03/18/2018] [Indexed: 05/23/2023]
Abstract
BACKGROUND Chemotherapy has been widely used to treat cancer, but the appearance of multidrug resistance (MDR) is the biggest obstacle to successful chemotherapy. One of the conventional mechanisms of MDR is overexpression of ATP-binding cassette (ABC) transporters such as P-glycoprotein (P-gp/ABCB1) and multidrug resistance-associated proteins (MRPs/ABCCs) that limits the prolonged and efficient use of chemotherapeutic drugs. To enhance the chemosensitivity of tumor cells, attentions have been focused on effective MDR modulators. PURPOSE This study aimed to investigate the reversal effect of quercetin on MDR, and explored its mechanism of action in vitro. STUDY DESIGN/METHODS The effect and mechanism of quercetin on MDR was examined by using MTT assay, flow cytometry, real-time PCR and western blot analysis in human hepatocellular carcinoma cells. RESULTS Our data found that the intracellular accumulation of rhodamine-123 (Rh123) and doxorubicin (ADR) were increased, the sensitivity of BEL/5-FU cells to chemotherapeutic drugs were increased, and the expressions of ABCB1, ABCC1 and ABCC2 were all down-regulated, which indicated that the functions and expressions of ABCB1, ABCC1 and ABCC2 efflux pump were inhibited by quercetin treatment. Moreover, the suppression of ABCB1, ABCC1 and ABCC2 by quercetin was dependent on the FZD7 through the Wnt/β-catenin pathway. Further research revealed that reduction of FZD7 by RNA interference (siFZD7) enhanced the sensitivity to chemotherapeutic drugs, increased the cellular accumulation of Rh123 and ADR, and induced inhibitory effects on the expression of FZD7, ABCB1, ABCC1, ABCC2 and β-catenin, similar to quercetin. In the meanwhile, overexpression of FZD7 showed the inversely effect on the expressions. Interesting, it was confirmed that quercetin could inhibit the expression levels of FZD7, ABCB1, ABCC1, ABCC2 and β-catenin in BEL-7402 cells; furthermore, treatment by quercetin combined with siFZD7 in BEL/5-FU cells, the expressions of these genes were effectively decreased in comparison to quercetin combined with siRNA negative control (sncRNA). CONCLUSION Overall, these data suggested the effectiveness of using quercetin, at least in part, via inhibiting FZD7 to combat chemoresistance and showed that quercetin could be developed into an efficient natural sensitizer for resistant human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhaolin Chen
- Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, China; Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Cheng Huang
- Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, China
| | - Taotao Ma
- Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, China
| | - Ling Jiang
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Liqin Tang
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Tianlu Shi
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Shantang Zhang
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Lei Zhang
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Pengli Zhu
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Jun Li
- Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Aizong Shen
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China.
| |
Collapse
|
112
|
Zhang M, Wang Z, Zhang Y, Guo W, Ji H. Structure-Based Optimization of Small-Molecule Inhibitors for the β-Catenin/B-Cell Lymphoma 9 Protein-Protein Interaction. J Med Chem 2018; 61:2989-3007. [PMID: 29566337 DOI: 10.1021/acs.jmedchem.8b00068] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Structure-based optimization was conducted to improve the potency, selectivity, and cell-based activities of β-catenin/B-cell lymphoma 9 (BCL9) inhibitors based on the 4'-fluoro- N-phenyl-[1,1'-biphenyl]-3-carboxamide scaffold, which was designed to mimic the side chains of the hydrophobic α-helical hot spots at positions i, i + 3, and i + 7. Compound 29 was found to disrupt the β-catenin/BCL9 protein-protein interaction (PPI) with a Ki of 0.47 μM and >1900-fold selectivity for β-catenin/BCL9 over β-catenin/E-cadherin PPIs. The proposed binding mode of new inhibitors was consistent with the results of site-directed mutagenesis and structure-activity relationship studies. Cell-based studies indicated that 29 disrupted the β-catenin/BCL9 interaction without affecting the β-catenin/E-cadherin interaction, selectively suppressed transactivation of Wnt/β-catenin signaling, downregulated expression of Wnt target genes, and inhibited viability of Wnt/β-catenin-dependent cancer cells in dose-dependent manners. A comparison of the biochemical and cell-based assay results offered the directions for future inhibitor optimization.
Collapse
Affiliation(s)
- Min Zhang
- Drug Discovery Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , Florida 33612 , United States.,Departments of Oncologic Sciences and Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Zhen Wang
- Drug Discovery Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , Florida 33612 , United States.,Departments of Oncologic Sciences and Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Yongqiang Zhang
- Drug Discovery Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , Florida 33612 , United States.,Departments of Oncologic Sciences and Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Wenxing Guo
- Drug Discovery Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , Florida 33612 , United States.,Departments of Oncologic Sciences and Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Haitao Ji
- Drug Discovery Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , Florida 33612 , United States.,Departments of Oncologic Sciences and Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| |
Collapse
|
113
|
Condello S, Sima L, Ivan C, Cardenas H, Schiltz G, Mishra RK, Matei D. Tissue Tranglutaminase Regulates Interactions between Ovarian Cancer Stem Cells and the Tumor Niche. Cancer Res 2018; 78:2990-3001. [PMID: 29510995 DOI: 10.1158/0008-5472.can-17-2319] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/15/2017] [Accepted: 03/02/2018] [Indexed: 12/12/2022]
Abstract
Cancer progression and recurrence are linked to a rare population of cancer stem cells (CSC). Here, we hypothesized that interactions with the extracellular matrix drive CSC proliferation and tumor-initiating capacity and investigated the functions of scaffold protein tissue transglutaminase (TG2) in ovarian CSC. Complexes formed by TG2, fibronectin (FN), and integrin β1 were enriched in ovarian CSC and detectable in tumors. A function-inhibiting antibody against the TG2 FN-binding domain suppressed complex formation, CSC proliferation as spheroids, tumor-initiating capacity, and stemness-associated Wnt/β-catenin signaling. Disruption of the interaction between TG2 and FN also blocked spheroid formation and the response to Wnt ligands. TG2 and the Wnt receptor Frizzled 7 (Fzd7) form a complex in cancer cells and tumors, leading to Wnt pathway activation. Protein docking and peptide inhibition demonstrate that the interaction between TG2 and Fzd7 overlaps with the FN-binding domain of TG2. These results support a new function of TG2 in ovarian CSC, linked to spheroid proliferation and tumor-initiating capacity and mediated through direct interactions with Fzd7. We propose this complex as a new stem cell target.Significance: These findings reveal a new mechanism by which ovarian CSCs interact with the tumor microenvironment, promoting cell proliferation and tumor initiation. Cancer Res; 78(11); 2990-3001. ©2018 AACR.
Collapse
Affiliation(s)
- Salvatore Condello
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Livia Sima
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Horacio Cardenas
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Gary Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois
| | - Rama K Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
- Robert H Lurie Comprehensive Cancer Center, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
114
|
Chalakur-Ramireddy NKR, Pakala SB. Combined drug therapeutic strategies for the effective treatment of Triple Negative Breast Cancer. Biosci Rep 2018; 38:BSR20171357. [PMID: 29298879 PMCID: PMC5789156 DOI: 10.1042/bsr20171357] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/12/2017] [Accepted: 01/01/2018] [Indexed: 12/19/2022] Open
Abstract
TNBC (Triple Negative Breast Cancer) is a subtype of breast cancer with an aggressive phenotype which shows high metastatic capability and poor prognosis. Owing to its intrinsic properties like heterogeneity, lack of hormonal receptors and aggressive phenotype leave chemotherapy as a mainstay for the treatment of TNBC. Various studies have demonstrated that chemotherapy alone or therapeutic drugs targeting TNBC pathways, epigenetic mechanisms and immunotherapy alone have not shown significant improvement in TNBC patients. On the other hand, a combination of therapeutic drugs or addition of chemotherapy with therapeutic drugs has shown substantial improvement in results and proven to be an effective strategy for TNBC treatment. This review sheds light on effective combinational drug strategies and current clinical trial status of various combinatorial drugs for the treatment of TNBC.
Collapse
Affiliation(s)
| | - Suresh B Pakala
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, Andhra Pradesh, India
| |
Collapse
|
115
|
Qiu X, Jiao J, Li Y, Tian T. Overexpression of FZD7 promotes glioma cell proliferation by upregulating TAZ. Oncotarget 2018; 7:85987-85999. [PMID: 27852064 PMCID: PMC5349891 DOI: 10.18632/oncotarget.13292] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/03/2016] [Indexed: 01/27/2023] Open
Abstract
Gliomas are the most prevalent type of primary brain tumors in adults, accounting for more than 40% of neoplasm in the central nervous system. Frizzled-7 (FZD7) is a seven-pass trans-membrane Wnt receptor that plays a critical role in the development of various tumors. In this study, we detected high-level FZD7 expression in glioma and its overexpression was associated with advanced tumor stage. In vitro functional assays showed that forced overexpression of FZD7 promoted proliferation of gliomas cells, whereas knockdown of endogenous FZD7 significantly suppressed proliferation ability of these cells. In a xenograft assay, FZD7 was also found to promote the growth of glioma cells. We further found that FZD7 could activate transcriptional coactivator with PDZ-binding motif (TAZ), and TAZ was required for FZD7 to promote cell proliferation in glioma. Furthermore, the univariate analysis of survival shows that glioma patients with high FZD7 expression have a shorter survival. In conclusion, our findings demonstrate that FZD7 may promote glioma cell proliferation via upregulation of TAZ.
Collapse
Affiliation(s)
- Xia Qiu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Department of Medicine, Shangqiu Medical School, Shangqiu, Henan Province, China
| | - Jianguo Jiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yidong Li
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Tian Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
116
|
Li G, Su Q, Liu H, Wang D, Zhang W, Lu Z, Chen Y, Huang X, Li W, Zhang C, He Y, Fu L, Bi J. Frizzled7 Promotes Epithelial-to-mesenchymal Transition and Stemness Via Activating Canonical Wnt/β-catenin Pathway in Gastric Cancer. Int J Biol Sci 2018; 14:280-293. [PMID: 29559846 PMCID: PMC5859474 DOI: 10.7150/ijbs.23756] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/06/2018] [Indexed: 12/25/2022] Open
Abstract
Aberrant activation of Wnt signaling is a crucial event in tumor development and metastasis. Wnt signaling is commonly divided into canonical and non-canonical signaling pathways based on whether β-catenin is activated (canonical). The two signaling pathways are initiated by Wnt ligand binding to the surface Frizzled (FZD) receptors, and regulate cancer stem cell self-renewal and epithelial-mesenchymal transition (EMT). Frizzled 7 (FZD7), a member of Frizzled family, promotes cell proliferation and invasiveness in many cancers, suggesting that FZD7 transmitting Wnt signaling is important for driving cancer growth. FZD7 expression has been reported to be up-regulated in human primary gastric cancer tissues. However, the molecular mechanism by which FZD7 promotes gastric cancer(GC) development and progression is not fully understood. Our present study showed that FZD7 was overexpressed in clinical GC samples, and thus was correlated with tumor invasion, lymphatic and organ metastasis, late TNM stages and poor patient survival. The endogenous expression of FZD7 was significantly increased in cancer stem cell-enriched spheres compared with adherent cells. Furthermore, RNA interference-mediated silencing of FZD7 inhibited proliferation, migration and invasion in gastric cancer cells. Moreover, ablation of FZD7 down-regulated EMT and the expression levels of cancer stem cell markers, and these inhibitions were associated with attenuated canonical Wnt/β-catenin signaling. The results suggest that Wnt canonical pathway may contribute to tumorigenesis and metastasis, indicating that FZD7 could be a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Guanman Li
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Qiao Su
- Animal Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Haibo Liu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510000, Guangdong, China
| | - Dong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Wenhui Zhang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Yu Chen
- Department of Immunity, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Xiaohui Huang
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Wen Li
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Changhua Zhang
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yulong He
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Li Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Cancer Research Centre, School of Medicine, Shenzhen University, Shenzhen, China
| | - Jiong Bi
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
117
|
Zarei N, Fazeli M, Mohammadi M, Nejatollahi F. Cell growth inhibition and apoptosis in breast cancer cells induced by anti-FZD7 scFvs: involvement of bioinformatics-based design of novel epitopes. Breast Cancer Res Treat 2018; 169:427-436. [DOI: 10.1007/s10549-017-4641-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/23/2017] [Indexed: 02/06/2023]
|
118
|
Yin L, Gao Y, Zhang X, Wang J, Ding D, Zhang Y, Zhang J, Chen H. Niclosamide sensitizes triple-negative breast cancer cells to ionizing radiation in association with the inhibition of Wnt/β-catenin signaling. Oncotarget 2018; 7:42126-42138. [PMID: 27363012 PMCID: PMC5173121 DOI: 10.18632/oncotarget.9704] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/16/2016] [Indexed: 01/22/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most difficult breast cancers to treat because there is no targeted treatment, and conventional cytotoxic chemotherapy followed by adjuvant radiation therapy is the standard of care for patients with TNBC. We herein reported that ionizing radiation (IR) induced Wnt3a, LRP6 and β-catenin expression and consequently activated Wnt/β-catenin signaling in TNBC MDA-MB-231, MDA-MB-468 and Hs578T cells. Moreover, depletion of β-catenin by shRNA sensitized TNBC cells to IR, whereas treatment of Wnt3a protein or overexpression of β-catenin resulted in radioresistance of TNBC cells. Niclosamide, a potent inhibitor of Wnt/β-catenin signaling, not only inhibited constitutive Wnt/β-catenin signaling, but also blocked IR-induced Wnt/β-catenin signaling in TNBC cells. In addition, niclosamide sensitized TNBC cells to IR, prevented Wnt3a-induced radioresistance, and overcame β-catenin-induced radioresistance in TNBC cells. Importantly, animals treated with the combination of niclosamide and γ-ray local tumor irradiation had significant inhibition of MDA-MB-231 tumor growth compared with treated with local tumor irradiation alone. These findings indicate that Wnt/β-catenin signaling pathway plays an important role in the development of radioresistance of TNBC cells, and that niclosamide had significant radiosensitizing effects by inhibiting Wnt/β-catenin signaling in TNBC cells. Our study also provides rationale for further preclinical and clinical evaluation of niclosamide in TNBC management.
Collapse
Affiliation(s)
- Lina Yin
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Yun Gao
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Xuxia Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Jing Wang
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Defang Ding
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Yaping Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Junxiang Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Honghong Chen
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, China
| |
Collapse
|
119
|
Sriroopreddy R, Sudandiradoss C. Integrative network-based approach identifies central genetic and transcriptomic elements in triple-negative breast cancer. Funct Integr Genomics 2017; 18:113-124. [DOI: 10.1007/s10142-017-0579-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/18/2022]
|
120
|
Guo L, Wang X, Yang Y, Xu H, Zhang Z, Yin L, Wang Y, Yang M, Zhao S, Bai S, Zhao L, Wang Z, Lian X, Liu Y, Zhang Q. Methylation of DACT2 contributes to the progression of breast cancer through activating WNT signaling pathway. Oncol Lett 2017; 15:3287-3294. [PMID: 29435071 DOI: 10.3892/ol.2017.7633] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/11/2016] [Indexed: 12/16/2022] Open
Abstract
The activation of the Wnt/β-catenin signaling pathway has been demonstrated to play important roles in breast carcinogenesis and to be associated with a poorer prognosis in breast cancer patients. However, genetic mutation is not the major reason for Wnt/β-catenin activation in breast cancer. Dishevelled-associated antagonist of β-catenin homolog 2 (DACT2) is a negative regulator of β-catenin and acts as a tumor suppressor in numerous cancer types; however, the expression change and potential role of DACT2 in breast cancer is unknown. The present study detected the expression and function of DACT2 in breast cancer progression. It was identified that the expression of DACT2 significantly decreased in breast cancer tissues compared with paired adjacent normal breast tissues. Additional investigation demonstrated that the hypermethylation of DACT2 gene promoter contributes to the loss of the gene in breast cancer. It was also demonstrated that DACT2 is a tumor suppressor in breast cancer and inhibits the proliferation and invasion of breast cancer cells by repressing the expression of β-catenin target genes associated with tumor growth and metastasis. The present study indicates that the loss of DACT2 may contribute to breast cancer progression and provides a promising therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Li Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaohong Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Yuguang Yang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Hongchun Xu
- Department of Thoracic Surgery, Mudanjiang Tumor Hospital, Mudanjiang, Heilongjiang 157000, P.R. China
| | - Zhihong Zhang
- Obstetrics and Gynecology Department, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163000, P.R. China
| | - Lili Yin
- Obstetrics and Gynecology Department, Heilongjiang Electric Power Hospital, Harbin, Heilongjiang 150090, P.R. China
| | - Yan Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Maopeng Yang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Shu Zhao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Shuping Bai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Ling Zhao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Zhipeng Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Xin Lian
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Ying Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
121
|
Driehuis E, Clevers H. WNT signalling events near the cell membrane and their pharmacological targeting for the treatment of cancer. Br J Pharmacol 2017; 174:4547-4563. [PMID: 28244067 PMCID: PMC5727251 DOI: 10.1111/bph.13758] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 12/16/2022] Open
Abstract
WNT signalling is an essential signalling pathway for all multicellular animals. Although first described more than 30 years ago, new components and regulators of the pathway are still being discovered. Considering its importance in both embryonic development and adult homeostasis, it is not surprising that this pathway is often deregulated in human diseases such as cancer. Recently, it became clear that in addition to cytoplasmic components such as β-catenin, other, membrane-bound or extracellular, components of the WNT pathway are also altered in cancer. This review gives an overview of the recent discoveries on WNT signalling events near the cell membrane. Furthermore, membrane-associated components of the WNT pathway, which are more accessible for therapeutic intervention, as well therapeutic approaches that already target those components will be discussed. In this way, we hope to stimulate the development of effective anti-cancer therapies that target this fascinating pathway. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Else Driehuis
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW)UtrechtThe Netherlands
- University medical center (UMC)UtrechtThe Netherlands
| | - Hans Clevers
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW)UtrechtThe Netherlands
- University medical center (UMC)UtrechtThe Netherlands
- Princess Maxime Center (PMC)UtrechtThe Netherlands
| |
Collapse
|
122
|
High-throughput targeted screening in triple-negative breast cancer cells identifies Wnt-inhibiting activities in Pacific brittle stars. Sci Rep 2017; 7:11964. [PMID: 28931883 PMCID: PMC5607299 DOI: 10.1038/s41598-017-12232-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/06/2017] [Indexed: 02/06/2023] Open
Abstract
Pro-proliferative oncogenic signaling is one of the hallmarks of cancer. Specific targeting of such signaling pathways is one of the main approaches to modern anti-cancer drug discovery, as opposed to more traditional search for general cytotoxic agents. Natural products, especially from marine sources, represent a largely untapped source of chemical diversity, which so far have mostly been screened for cytotoxicity. Here we present a pioneering pipeline of high-throughput screening of marine-based activities targeted against the Wnt signaling pathway, which is one of the key factors in oncogenic transformation, growth and metastasis in different cancers, including the devastating triple-negative breast cancer (TNBC) currently lacking any targeted therapies. This pipeline consisted of collection and characterization of numerous invertebrates during the SokhoBio expedition to the Kuril Basin in North Pacific, preparation of extracts from these specimen, and their screening in dedicated assays monitoring Wnt signaling in TNBC cells. This approach yielded a number of promising hits, including highly specific anti-Wnt activities targeting multiple levels within the Wnt pathway from Ophiura irrorata and other Pacific brittle stars.
Collapse
|
123
|
Zhang W, Lu W, Ananthan S, Suto MJ, Li Y. Discovery of novel frizzled-7 inhibitors by targeting the receptor's transmembrane domain. Oncotarget 2017; 8:91459-91470. [PMID: 29207657 PMCID: PMC5710937 DOI: 10.18632/oncotarget.20665] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023] Open
Abstract
Frizzled (Fzd) proteins are seven transmembrane receptors that belong to a novel and separated family of G-protein-coupled receptors (GPCRs). The Fzd receptors can respond to Wnt proteins to activate the canonical β-catenin pathway which is important for both initiation and progression of cancers. Disruption of the Wnt/β-catenin signal thus represents an opportunity for rational cancer prevention and therapy. Of the 10 members of the Fzd family, Fzd7 is the most important member involved in cancer development and progression. In the present studies, we applied structure-based virtual screening targeting the transmembrane domain (TMD) of Fzd7 to select compounds that could potentially bind to the Fzd7-TMD and block the Wnt/Fzd7 signaling and further evaluated them in biological assays. Six small molecule compounds were confirmed as Fzd7 inhibitors. The best hit, SRI37892, significantly blocked the Wnt/Fzd7 signaling with IC50 values in the sub-micromolar range and inhibited cancer cell proliferation with IC50 values around 2 μM. Our results provide the first proof of concept of targeting Fzd-TMD for the development of Wnt/Fzd modulators. The identified small molecular Fzd7 inhibitors can serve as a useful tool for studying the regulation mechanism(s) of Wnt/Fzd7 signaling as well as a starting point for the development of cancer therapeutic agents.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States
| | - Wenyan Lu
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States
| | - Subramaniam Ananthan
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States
| | - Mark J Suto
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States
| | - Yonghe Li
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States
| |
Collapse
|
124
|
Ma J, Lu W, Chen D, Xu B, Li Y. Role of Wnt Co-Receptor LRP6 in Triple Negative Breast Cancer Cell Migration and Invasion. J Cell Biochem 2017; 118:2968-2976. [PMID: 28247948 PMCID: PMC10928515 DOI: 10.1002/jcb.25956] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/27/2017] [Indexed: 03/14/2024]
Abstract
The low-density lipoprotein receptor-related protein 6 (LRP6) is an essential Wnt co-receptor of the Wnt/β-catenin signaling pathway. Although studies have shown an increased expression of LRP6 in several types of cancer, its function in tumor development and progression remains to be elucidated. We herein demonstrated that LRP6 expression is up-regulated in human triple negative breast cancer (TNBC) patients and human TNBC cell lines, and that knockdown of LRP6 expression and treatment of recombinant Mesd protein (a specific inhibitor of LRP6) significantly decreased cell migration and invasion of TNBC MDA-MB-231 and BT549 cells. Interestingly, the effects of LRP6 knockdown and Mesd treatment on TNBC cell migration and invasion were more prominent than on TNBC cell proliferation/viability. Mechanistically, LRP6 knockdown and Mesd treatment inhibited Wnt/β-catenin signaling and decreased the expression of S100A4, a mediator of cancer metastasis and a specific target of Wnt/β-catenin signaling, in TNBC cells. Together, our data suggest that LRP6 promotes TNBC cell migration and invasion by regulating the expression and function of S100A4 via the Wnt/β-catenin signaling pathway. J. Cell. Biochem. 118: 2968-2976, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jinlu Ma
- Department of Radiation Oncology, the First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
- Department of Oncology, Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35255, USA
| | - Wenyan Lu
- Department of Oncology, Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35255, USA
| | - Dongquan Chen
- Division of Preventive Medicine and Comprehensive Cancer Center; Department of Medicine; University of Alabama at Birmingham; Birmingham, AL USA
| | - Bo Xu
- Department of Oncology, Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35255, USA
| | - Yonghe Li
- Department of Oncology, Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35255, USA
| |
Collapse
|
125
|
Guo J, Gong G, Zhang B. Screening and identification of potential biomarkers in triple-negative breast cancer by integrated analysis. Oncol Rep 2017; 38:2219-2228. [PMID: 28849078 DOI: 10.3892/or.2017.5911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/29/2017] [Indexed: 11/06/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has attracted great attention due to its unique biology, poor prognosis, and aggressiveness. TNBC patients are more likely to suffer from metastasis. We screened and identified the TNBC-specific genes as potential biomarkers. A total of 167 breast cancer samples (45 TNBC and 122 non-TNBC) were used in the integrated analysis. Gene expression microarrays were used to screen the differentially expressed genes. We identified 65 core DEGs. According to the GO and KEGG analysis, the gene function enrichment in TNBC was revealed, such as basal cell carcinoma, prostate cancer, oocyte meiosis and choline metabolism in cancer pathways. Moreover, the PPI network reconstruction would benefit the screening of hubs. A RFS analysis of TNBC-specific genes was also conducted. RT-PCR was used to validate the expression pattern of hubs in TNBC. Finally, nine genes were identified and all of them were novel, specific and higher dysregulation expressed genes in TNBC. Such that, these genes will serve as potential biomarkers in TNBC and benefit further research in TNBC.
Collapse
Affiliation(s)
- Jilong Guo
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| | - Guohua Gong
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| | - Bin Zhang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| |
Collapse
|
126
|
Shao F, Sun H, Deng CX. Potential therapeutic targets of triple-negative breast cancer based on its intrinsic subtype. Oncotarget 2017; 8:73329-73344. [PMID: 29069872 PMCID: PMC5641215 DOI: 10.18632/oncotarget.20274] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/06/2017] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subgroup of human breast cancer, which is characterized as estrogen receptor (ER) negative, progesterone receptor (PR) negative, and human epidermal growth factor receptor 2 (HER2) negative. TNBC is the most difficult breast cancer subgroup to treat, due to its unresponsiveness to current clinical targeted therapies, high rate of recurrence, and poor prognosis. Thus, there is an urgent medical need to identify therapeutic targets and develop more effective stratified medicine for the treatment of TNBC. Here we review the potential therapeutic targets for TNBC based on its intrinsic subtype. We also review the aberrant activated signals found in different subgroups of TNBC, including androgen receptor (AR) and PI3K/AKT/mTOR, Notch, Wnt/β-catenin, Hedge-hog, and TGF-β signaling pathways, which play essential roles in multiple development stages of TNBC. The careful analysis of these signaling pathways and therapeutic targets would have significant impact on the drug development and clinical trials, leading to effective therapies for this deadly disease.
Collapse
Affiliation(s)
- Fangyuan Shao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Heng Sun
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
127
|
Liu X, Yan Y, Ma W, Wu S. Knockdown of frizzled-7 inhibits cell growth and metastasis and promotes chemosensitivity of esophageal squamous cell carcinoma cells by inhibiting Wnt signaling. Biochem Biophys Res Commun 2017; 490:1112-1118. [DOI: 10.1016/j.bbrc.2017.06.185] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022]
|
128
|
Song H, Nan Y, Wang X, Zhang G, Zong S, Kong X. MicroRNA‑613 inhibits proliferation and invasion of renal cell carcinoma cells through targeting FZD7. Mol Med Rep 2017; 16:4279-4286. [PMID: 29067457 DOI: 10.3892/mmr.2017.7076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 05/18/2017] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as critical regulators in cancer progression. miR‑613 has been reported as a tumor suppressor gene in many types of human cancers. However, the function of miR‑613 in renal cell carcinoma (RCC) remains unclear. In the present study, the authors aimed to detect the expression of miR‑613 and its function in RCC cell lines. miR‑613 was reported to be significantly downregulated RCC cell lines. Functional analyses demonstrated that overexpression of miR‑613 significantly decreased RCC cell proliferation and invasion. Bioinformatics analysis showed that Frizzled7 (FZD7) was a predicted target of miR‑613, which was verified by dual‑luciferase reporter assay, reverse transcription quantitative‑polymerase chain reaction and western blot analysis. Restoration of FZD7 significantly reversed the suppressive effects of miR‑613 on RCC cell proliferation and invasion. Taken together, the results of the present study indicated that miR‑613 functions as a tumor suppressor that inhibits RCC cell proliferation and invasion by targeting and inhibiting FZD7, providing novel insight into RCC pathogenesis and a potential therapeutic target for RCC.
Collapse
Affiliation(s)
- Haitao Song
- Department of Urinary Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yonghao Nan
- Department of Urinary Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xinsheng Wang
- Department of Urinary Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Gang Zhang
- Department of Urinary Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Shi Zong
- Department of Urinary Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xiangbo Kong
- Department of Urinary Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
129
|
Polosukhina D, Love HD, Moses HL, Lee E, Zent R, Clark PE. Pharmacologic Inhibition of β-Catenin With Pyrvinium Inhibits Murine and Human Models of Wilms Tumor. Oncol Res 2017; 25:1653-1664. [PMID: 28695795 PMCID: PMC5670010 DOI: 10.3727/096504017x14992942781895] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Wilms tumor (WT) is the most common renal malignancy in children and the fourth most common pediatric solid malignancy in the US. Although the mechanisms underlying the WT biology are complex, these tumors most often demonstrate activation of the canonical Wnt/β-catenin pathway. We and others have shown that constitutive activation of β-catenin restricted to the renal epithelium is sufficient to induce primitive renal epithelial tumors, which resemble human WT. Here we demonstrate that pharmacologic inhibition of β-catenin gene transcription with pyrvinium inhibits tumor growth and metastatic progression in a murine model of WT. Cellular invasion is significantly inhibited in both murine WT-like and human WT cells and is accompanied by downregulation of the oncogenes Myc and Birc5 (survivin). Our studies provide proof of the concept that the canonical Wnt/β-catenin pathway may be a novel therapeutic target in the management of WT.
Collapse
|
130
|
Zhang S, Zhang Y, Yu P, Hu Y, Zhou H, Guo L, Xu X, Zhu X, Waqas M, Qi J, Zhang X, Liu Y, Chen F, Tang M, Qian X, Shi H, Gao X, Chai R. Characterization of Lgr5+ Progenitor Cell Transcriptomes after Neomycin Injury in the Neonatal Mouse Cochlea. Front Mol Neurosci 2017; 10:213. [PMID: 28725177 PMCID: PMC5496572 DOI: 10.3389/fnmol.2017.00213] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/16/2017] [Indexed: 12/17/2022] Open
Abstract
Lgr5+ supporting cells (SCs) are enriched hair cell (HC) progenitors in the cochlea. Both in vitro and in vivo studies have shown that HC injury can spontaneously activate Lgr5+ progenitors to regenerate HCs in the neonatal mouse cochlea. Promoting HC regeneration requires the understanding of the mechanism of HC regeneration, and this requires knowledge of the key genes involved in HC injury-induced self-repair responses that promote the proliferation and differentiation of Lgr5+ progenitors. Here, as expected, we found that neomycin-treated Lgr5+ progenitors (NLPs) had significantly greater HC regeneration ability, and greater but not significant proliferation ability compared to untreated Lgr5+ progenitors (ULPs) in response to neomycin exposure. Next, we used RNA-seq analysis to determine the differences in the gene-expression profiles between the transcriptomes of NLPs and ULPs from the neonatal mouse cochlea. We first analyzed the genes that were enriched and differentially expressed in NLPs and ULPs and then analyzed the cell cycle genes, the transcription factors, and the signaling pathway genes that might regulate the proliferation and differentiation of Lgr5+ progenitors. We found 9 cell cycle genes, 88 transcription factors, 8 microRNAs, and 16 cell-signaling pathway genes that were significantly upregulated or downregulated after neomycin injury in NLPs. Lastly, we constructed a protein-protein interaction network to show the interaction and connections of genes that are differentially expressed in NLPs and ULPs. This study has identified the genes that might regulate the proliferation and HC regeneration of Lgr5+ progenitors after neomycin injury, and investigations into the roles and mechanisms of these genes in the cochlea should be performed in the future to identify potential therapeutic targets for HC regeneration.
Collapse
Affiliation(s)
- Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Yuan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Pengfei Yu
- Bioinformatics Department, Admera Health LLCSouth Plainfield, NJ, United States
| | - Yao Hu
- School of Pharmacy, Institute for Stem Cell and Neural Regeneration, Nanjing Medical UniversityNanjing, China
| | - Han Zhou
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Lingna Guo
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaochen Xu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaocheng Zhu
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and TechnologyKarachi, Pakistan
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaoli Zhang
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Yan Liu
- School of Pharmacy, Institute for Stem Cell and Neural Regeneration, Nanjing Medical UniversityNanjing, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and TechnologyShenzhen, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaoyun Qian
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong UniversityShanghai, China
| | - Xia Gao
- Research Institute of OtolaryngologyNanjing, China.,Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| |
Collapse
|
131
|
Riley RS, Day ES. Frizzled7 Antibody-Functionalized Nanoshells Enable Multivalent Binding for Wnt Signaling Inhibition in Triple Negative Breast Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:10.1002/smll.201700544. [PMID: 28544579 PMCID: PMC5545881 DOI: 10.1002/smll.201700544] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/26/2017] [Indexed: 05/08/2023]
Abstract
Antibodies that antagonize cell signaling pathways specific to their targeted receptor are invaluable tools to study and treat malignancies, but their utility is limited by high production costs and treatment dosages. Researchers have shown that antibodies conjugated to nanoparticles display increased affinity for their target relative to freely delivered antibodies due to multivalency, and this study investigates how this multivalency can enable antibody-nanoparticle conjugates to inhibit oncogenic cell signaling more effectively than freely delivered antibodies. This effect was evaluated using triple negative breast cancer (TNBC) cells that are characterized by hyperactive Wnt signaling mediated through overexpressed Frizzled7 (FZD7) transmembrane receptors. Through analysis of the expression of β-catenin and Axin2, two downstream targets in the Wnt pathway, the results demonstrate that FZD7 antibody-nanoshell conjugates (FZD7-NS) are drastically more effective at inhibiting Wnt signaling in TNBC cells than freely delivered FZD7 antibodies. Additionally, cells treated with FZD7-NS, but not cells treated with freely delivered FZD7 antibodies, have decreased viability, indicating the therapeutic potential of this technology. The results demonstrate that antibody-functionalized nanoparticles can exploit multivalency for improved signal cascade interference over free antibodies, and this may ultimately permit lower antibody dosages to be administered to study signaling pathways or to manage diseases.
Collapse
Affiliation(s)
- Rachel S Riley
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, 150 Academy Street, Newark, DE, 19716, USA
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, 150 Academy Street, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, 19713, USA
| |
Collapse
|
132
|
Wu J, Li J, Ren J, Zhang D. MicroRNA-485-5p represses melanoma cell invasion and proliferation by suppressing Frizzled7. Biomed Pharmacother 2017; 90:303-310. [DOI: 10.1016/j.biopha.2017.03.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
|
133
|
Schmucker HS, Park JP, Coissieux MM, Bentires-Alj M, Feltus FA, Booth BW. RNA Expression Profiling Reveals Differentially Regulated Growth Factor and Receptor Expression in Redirected Cancer Cells. Stem Cells Dev 2017; 26:646-655. [DOI: 10.1089/scd.2016.0340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Hannah S. Schmucker
- Department of Biological Sciences, Clemson University, Clemson, South Carolina
| | - Jang Pyo Park
- Institute for Biological Interfaces of Engineering, Clemson University, Clemson, South Carolina
| | - Marie-May Coissieux
- Friedrich-Miescher Institute for Biomedical Research, Basel, Switzerland
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Friedrich-Miescher Institute for Biomedical Research, Basel, Switzerland
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - F. Alex Feltus
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina
| | - Brian W. Booth
- Institute for Biological Interfaces of Engineering, Clemson University, Clemson, South Carolina
- Department of Bioengineering, Clemson University, Clemson, South Carolina
| |
Collapse
|
134
|
Wnt signaling in triple-negative breast cancer. Oncogenesis 2017; 6:e310. [PMID: 28368389 PMCID: PMC5520491 DOI: 10.1038/oncsis.2017.14] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/09/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling regulates a variety of cellular processes, including cell fate, differentiation, proliferation and stem cell pluripotency. Aberrant Wnt signaling is a hallmark of many cancers. An aggressive subtype of breast cancer, known as triple-negative breast cancer (TNBC), demonstrates dysregulation in canonical and non-canonical Wnt signaling. In this review, we summarize regulators of canonical and non-canonical Wnt signaling, as well as Wnt signaling dysfunction that mediates the progression of TNBC. We review the complex molecular nature of TNBC and the emerging therapies that are currently under investigation for the treatment of this disease.
Collapse
|
135
|
Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene 2017; 36:1461-1473. [PMID: 27617575 PMCID: PMC5357762 DOI: 10.1038/onc.2016.304] [Citation(s) in RCA: 1862] [Impact Index Per Article: 232.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/07/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022]
Abstract
Wnt signaling is one of the key cascades regulating development and stemness, and has also been tightly associated with cancer. The role of Wnt signaling in carcinogenesis has most prominently been described for colorectal cancer, but aberrant Wnt signaling is observed in many more cancer entities. Here, we review current insights into novel components of Wnt pathways and describe their impact on cancer development. Furthermore, we highlight expanding functions of Wnt signaling for both solid and liquid tumors. We also describe current findings how Wnt signaling affects maintenance of cancer stem cells, metastasis and immune control. Finally, we provide an overview of current strategies to antagonize Wnt signaling in cancer and challenges that are associated with such approaches.
Collapse
Affiliation(s)
- T Zhan
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
- Heidelberg University, Department of Internal Medicine II, Medical Faculty Mannheim, Mannheim, Germany
| | - N Rindtorff
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
| | - M Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
136
|
Anti-Proliferative Effects of Human Anti-FZD7 Single Chain Antibodies on Colorectal Cancer Cells. ACTA ACUST UNITED AC 2017. [DOI: 10.5812/semj.45219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
137
|
Lin ZJ, Ming J, Yang L, Du JZ, Wang N, Luo HJ. Mechanism of Regulatory Effect of MicroRNA-206 on Connexin 43 in Distant Metastasis of Breast Cancer. Chin Med J (Engl) 2017; 129:424-34. [PMID: 26879016 PMCID: PMC4800843 DOI: 10.4103/0366-6999.176071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background: MicroRNA-206 (miR-206) and connexin 43 (Cx43) are related with the distant metastasis of breast cancer. It remains unclear whether the regulatory effect of miR-206 on Cx43 is involved in metastasis of breast cancer. Methods: Using quantitative real-time polymerase chain reaction and Western blot, the expressions of miR-206 and Cx43 were determined in breast cancer tissues, hepatic and pulmonary metastasis (PM), and cell lines (MCF-10A, MCF-7, and MDA-MB-231). MCF-7/MDA-M-231 cells were transfected with lentivirus-shRNA vectors to enhance/inhibit miR-206, and then Cx43 expression was observed. Cell counting kit-8 assay and Transwell method were used to detect their changes in proliferation, migration, and invasion activity. The mutant plasmids of Cx43-3’ untranslated region (3’UTR) at position 478–484 and position 1609–1615 were constructed. Luciferase reporter assay was performed to observe the effects of miR-206 on luciferase expression of different mutant plasmids and to confirm the potential binding sites of Cx43. Results: Cx43 protein expression in hepatic and PM was significantly higher than that in the primary tumor, while no significant difference was showed in messenger RNA (mRNA) expression. MiR-206 mRNA expression in hepatic and PM was significantly lower than that in the primary tumor. Cx43 mRNA and protein levels, as well as cell proliferation, migration, and invasion capabilities, were all significantly improved in MDA-MB-231 cells after reducing miR-206 expression but decreased in MCF-7 cells after elevating miR-206 expression, which demonstrated a significantly negative correlation between miR-206 and Cx43 expression (P = 0.03). MiR-206 can drastically decrease Cx43 expression of MCF-7 cells but exerts no effects on Cx43 expression in 293 cells transfected with the Cx43 coding region but the lack of Cx43-3’UTR, suggesting that Cx43-3’UTR may be the key in Cx43 regulated by miR-206. Luciferase expression showed that the inhibition efficiency was reduced by 46.80% in position 478–484 mutant, 16.72% in position 1609–1615 mutant; the inhibition was totally disappeared in double mutant (P = 0.02). Conclusions: MiR-206 can regulate the expression of Cx43, the cytobiological activity, and the metastasis of breast cancer through binding to the two binding sites in Cx43-3’UTR: position 478–484 and position 1609–1615.
Collapse
Affiliation(s)
| | - Jia Ming
- Department of Breast, Thyroid and Pancreas Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | | | | | | | | |
Collapse
|
138
|
Daulat AM, Borg JP. Wnt/Planar Cell Polarity Signaling: New Opportunities for Cancer Treatment. Trends Cancer 2017; 3:113-125. [PMID: 28718442 DOI: 10.1016/j.trecan.2017.01.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/29/2016] [Accepted: 01/02/2017] [Indexed: 01/08/2023]
Abstract
Cancer cells are addicted to a large spectrum of extracellular cues implicated in initiation, stem cell renewal, tumor growth, dissemination in the body, and resistance to treatment. Wingless/Int-1 (Wnt) ligands and their associated signaling cascades contribute to most of these processes, paving the way for opportunities in therapeutic development. The developmental Wnt/planar cell polarity (PCP) pathway is the most recently described branch of Wnt signaling strongly implicated in cancer development at early and late stages. We describe here some of the latest knowledge accumulated on this pathway and the pending questions, present the most convincing findings about its role in cancer, and review the most promising strategies currently designed to target its components.
Collapse
Affiliation(s)
- Avais M Daulat
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Inst Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, 'Cell Polarity, Cell Signalling, and Cancer - Equipe Labellisée Ligue Contre le Cancer', Marseille, France
| | - Jean-Paul Borg
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Inst Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, 'Cell Polarity, Cell Signalling, and Cancer - Equipe Labellisée Ligue Contre le Cancer', Marseille, France.
| |
Collapse
|
139
|
Abstract
Inspite of effective treatment with imatinib (IM), chronic myeloid leukemia (CML) is still an incurable disease. Some patients became refractory because of IM resistance. Bone marrow mesenchymal stem cells (BMSCs) have been implicated a role in promoting CML cells' resistance against IM treatment. The detailed molecular mechanisms, however, remain largely unknown. In this study, we found that BMSCs increased the expression of FZD7 and activated Wnt/β-catenin signaling pathway in CML cells. BMSCs from CML patients showed increased efficiency to accelerate CML cell proliferation, enhance the drug resistance of K562 cells and up-regulate the expression of FZD7. Antagonism of FZD7 expression by shRNA significantly suppressed proliferation and increased IM sensitivity of CML cells co-cultured with BMSCs cells. Our findings suggest that FZD7, involved in canonical Wnt signaling pathway, plays a critical role in mediating BMSCs-dependent protection of CML cells, and potentially provide a novel therapeutic target for CML.
Collapse
|
140
|
Poloz Y, Dowling RJO, Stambolic V. Fundamental Pathways in Breast Cancer 1: Signaling from the Membrane. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
141
|
Rismani E, Fazeli MS, Mahmoodzadeh H, Movassagh A, Azami S, Karimipoor M, Teimoori-Toolabi L. Pattern of LRP6 gene expression in tumoral tissues of colorectal cancer. Cancer Biomark 2017; 19:151-159. [PMID: 28387660 DOI: 10.3233/cbm-160175] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is known as one of the most leading causes of death from cancer in the world. Wnt signaling pathway is the most important signaling pathway activated in this type of cancer. The low-density lipoprotein (LDL) receptor-related protein-6 (LRP6) functions as a co-receptor in order to trigger this signaling pathway. OBJECTIVE The expression of LRP6 was studied in cell lines, tumoral and normal adjacent tissues from patients affected with colorectal cancer. MATERIALS AND METHODS The expression of LRP6 was assayed by quantitative Real time PCR. RESULTS LRP6 is overexpressed at the transcript and protein level in HCT116 and SW480 in comparison to HUVEC as the non-cancerous cell. Furthermore, LRP6 was significantly up-regulated in 61% of human malignant tissues (p-value = 0.0008). In inherited type of disease, this upregulation was lower than other types of colorectal cancer, while in rectal cancer the overexpression was significantly higher (p-value = 0.039). Gene expression was not correlated with the age, gender and the stage of disease. CONCLUSION Due to the profound overexpression of LRP6 in sporadic and rectal types of cancer compared to normal colonic ones, antagonist related approaches can be promising for targeted therapies of cancer.
Collapse
Affiliation(s)
- Elham Rismani
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Sadegh Fazeli
- Department of Surgery, Division of Colo-Rectal Surgery, Imam Khomeini Medical Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Habibollah Mahmoodzadeh
- Cancer Institute of Iran, Imam Khomeini Medical Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Aghdas Movassagh
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Sina Azami
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | | | | |
Collapse
|
142
|
Wu W, Dang S, Feng Q, Liang J, Wang Y, Fan N. MicroRNA-542-3p inhibits the growth of hepatocellular carcinoma cells by targeting FZD7/Wnt signaling pathway. Biochem Biophys Res Commun 2017; 482:100-105. [PMID: 27815069 DOI: 10.1016/j.bbrc.2016.10.136] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/28/2016] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNA) are relevant regulators of the tumorigenesis of various cancers, such as hepatocellular carcinoma (HCC). Recent studies have suggested that miR-542-3p is a tumor suppressor gene in numerous cancers. However, the role of miR-542-3p in HCC remains unclear. This study showed that miR-542-3p was downregulated in HCC tissues and cell lines. MTT, colony formation, and cell cycle assays revealed that miR-542-3p overexpression inhibited HCC cell growth, whereas miR-542-3p suppression promoted cell growth. Frizzled7 (FZD7), the most important Wnt receptor involved in cancer development and progression, was identified as a functional target of miR-542-3p through dual-luciferase reporter assay, RT-qPCR, and Western blot. The mRNA expression of FZD7 was inversely correlated with miR-542-3p expression in HCC tissues. miR-542-3p overexpression could significantly decrease the activation of Wnt signaling pathway in HCC cells. FZD7 overexpression could significantly reverse the inhibitory effect of miR-542-3p on HCC cell growth and Wnt signaling pathway. Taken together, our study suggests that miR-542-3p inhibits HCC cell growth by targeting FZD7 and inhibiting Wnt signaling pathway. The decreased miR-542-3p expression may also contribute to the progression of HCC and may represent a novel molecular therapeutic target for HCC.
Collapse
Affiliation(s)
- Wenhua Wu
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shuangsuo Dang
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Qinhui Feng
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Junrong Liang
- Department of Gastroenterology Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yuan Wang
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Na Fan
- Department of Respiratory Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| |
Collapse
|
143
|
Corda G, Sala G, Lattanzio R, Iezzi M, Sallese M, Fragassi G, Lamolinara A, Mirza H, Barcaroli D, Ermler S, Silva E, Yasaei H, Newbold RF, Vagnarelli P, Mottolese M, Natali PG, Perracchio L, Quist J, Grigoriadis A, Marra P, Tutt AN, Piantelli M, Iacobelli S, De Laurenzi V, Sala A. Functional and prognostic significance of the genomic amplification of frizzled 6 (FZD6) in breast cancer. J Pathol 2016; 241:350-361. [PMID: 27859262 PMCID: PMC5248601 DOI: 10.1002/path.4841] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 09/09/2016] [Accepted: 10/18/2016] [Indexed: 12/15/2022]
Abstract
Frizzled receptors mediate Wnt ligand signalling, which is crucially involved in regulating tissue development and differentiation, and is often deregulated in cancer. In this study, we found that the gene encoding the Wnt receptor frizzled 6 (FZD6) is frequently amplified in breast cancer, with an increased incidence in the triple‐negative breast cancer (TNBC) subtype. Ablation of FZD6 expression in mammary cancer cell lines: (1) inhibited motility and invasion; (2) induced a more symmetrical shape of organoid three‐dimensional cultures; and (3) inhibited bone and liver metastasis in vivo. Mechanistically, FZD6 signalling is required for the assembly of the fibronectin matrix, interfering with the organization of the actin cytoskeleton. Ectopic delivery of fibronectin in FZD6‐depleted, triple‐negative MDA‐MB‐231 cells rearranged the actin cytoskeleton and restored epidermal growth factor‐mediated invasion. In patients with localized, lymph node‐negative (early) breast cancer, positivity of tumour cells for FZD6 protein identified patients with reduced distant relapse‐free survival. Multivariate analysis indicated an independent prognostic significance of FZD6 expression in TNBC tumours, predicting distant, but not local, relapse. We conclude that the FZD6–fibronectin actin axis identified in our study could be exploited for drug development in highly metastatic forms of breast cancer, such as TNBC. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gabriele Corda
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Gianluca Sala
- MediaPharma srl, Chieti, Italy.,Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Rossano Lattanzio
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Manuela Iezzi
- Dipartimento di Medicina e Scienze dell'Invecchiamento, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Michele Sallese
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy.,Fondazione Mario Negri Sud, S. Maria Imbaro, Italy
| | - Giorgia Fragassi
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy.,Fondazione Mario Negri Sud, S. Maria Imbaro, Italy
| | - Alessia Lamolinara
- Dipartimento di Medicina e Scienze dell'Invecchiamento, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Hasan Mirza
- Breast Cancer Now Unit, Research Oncology, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Daniela Barcaroli
- Dipartimento di Scienze Psicologiche, della Salute e del Territorio, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Sibylle Ermler
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Elisabete Silva
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Hemad Yasaei
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Robert F Newbold
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Paola Vagnarelli
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | | | | | | | - Jelmar Quist
- Breast Cancer Now Unit, Research Oncology, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Anita Grigoriadis
- Breast Cancer Now Unit, Research Oncology, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Pierfrancesco Marra
- Breast Cancer Now Unit, Research Oncology, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Andrew N Tutt
- Breast Cancer Now Unit, Research Oncology, King's Health Partners AHSC, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Breast Cancer Now, The Institute of Cancer Research, London, UK
| | - Mauro Piantelli
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Stefano Iacobelli
- MediaPharma srl, Chieti, Italy.,Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Vincenzo De Laurenzi
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| | - Arturo Sala
- College of Health and Life Sciences, Brunel University London, Uxbridge, UK.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK.,Dipartimento di Scienze Psicologiche, della Salute e del Territorio, CESI-MeT, University G. D'Annunzio, Chieti, Italy
| |
Collapse
|
144
|
γKlotho is a novel marker and cell survival factor in a subset of triple negative breast cancers. Oncotarget 2016; 7:2611-28. [PMID: 26556877 PMCID: PMC4823059 DOI: 10.18632/oncotarget.6006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/04/2015] [Indexed: 12/31/2022] Open
Abstract
Over the last decade, breast cancer mortality has declined. However, triple negative breast cancer (TNBC) remains a challenging problem mostly due to early recurrence and lack of molecularly driven treatments. There is a critical need to identify subgroups of TNBC with common molecular features that can be therapeutically targeted. Here we show that in contrast to Klotho and βKlotho, the third member of the Klotho protein family, γKlotho, is overexpressed in more than 60% of TNBCs and correlates with poorer disease progression. Furthermore, we find that γKlotho is expressed in a subset of TNBC cell lines promoting cell growth. Importantly, we demonstrate that in these cells γKlotho is necessary for cell survival and that its depletion leads to constitutive ERK activation, cell cycle arrest and apoptosis. Interestingly, we observe increased oxidative stress in γKlotho-depleted cells suggesting that γKlotho enables cancer cells to cope with an oxidative environment and that cells become dependent on its expression to maintain this survival advantage. These findings indicate that γKlotho might be a potential marker for patients that would benefit from treatments that alter oxidative stress and constitutes a novel drug target for a subset of TN breast cancers.
Collapse
|
145
|
Nickho H, Younesi V, Aghebati-Maleki L, Motallebnezhad M, Majidi Zolbanin J, Movassagh Pour A, Yousefi M. Developing and characterization of single chain variable fragment (scFv) antibody against frizzled 7 (Fzd7) receptor. Bioengineered 2016; 8:501-510. [PMID: 27849134 DOI: 10.1080/21655979.2016.1255383] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
ABSTACT Wnt/β-catenin signaling pathway through Frizzled receptors has been shown to play a key role in both normal development and tumorigenesis. Overexpression of Wnt pathway genes, such as Fzd7 in several malignancies is well-documented. Therefore, targeting of Fzd7 and its ligand inhibits cancer cells proliferation metastasis. In the present study we isolated single chain variable fragments (scFvs) against Fzd7 receptor using phage display method. Semi-synthetic human naive antibody libraries (Tomlinson I + J) was employed in panning procedure to isolate specific scFv against specific peptide from extracellular domain of Fzd7 receptor. The reactivity and growth inhibition effects of the selected antibodies was evaluated using enzyme-linked immunosorbent assay (ELISA), MTT and annexin V assays, respectively. Seven scFvs reactive to Fzd7 were selected following 4 rounds of panning. The results showed that the selected scFvs inhibits cell growth through apoptosis cell death in a triple negative breast cancer cells, MDA-MB-231. Given that Fzd7 and Wnt pathway plays a critical role in tumor progression, selected blocking scFvs represent significant potential for immunotherapy of breast cancer cells.
Collapse
Affiliation(s)
- Hamid Nickho
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Vahid Younesi
- d Pishtaz Teb Zaman Diagnostics , Tehran , Iran.,e Department of Laboratory Sciences , Faculty of Paramedical Sciences, Alborz University of Medical Sciences , Karaj , Iran
| | - Leili Aghebati-Maleki
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Morteza Motallebnezhad
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Jafar Majidi Zolbanin
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Aliakbar Movassagh Pour
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mehdi Yousefi
- b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
146
|
Golenia G, Gatie MI, Kelly GM. Frizzled gene expression and negative regulation of canonical WNT-β-catenin signaling in mouse F9 teratocarcinoma cells. Biochem Cell Biol 2016; 95:251-262. [PMID: 28177772 DOI: 10.1139/bcb-2016-0150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mouse F9 cells differentiate into primitive endoderm (PrE) following the activation of the canonical WNT-β-catenin pathway. The upregulation of Wnt6 and activation of β-catenin-TCF-LEF-dependent transcription is known to accompany differentiation, but the Frizzled (FZD) receptor responsible for transducing the WNT6 signal is not known. Eight of the 10 Fzd genes were found to be expressed in F9 cells, with Fzd7 being the most highly expressed, and chosen for further analysis. To alter steady-state Fzd7 levels and test the effect this has on differentiation, siRNA and overexpression approaches were used to knock-down and ectopically express the Fzd7 message, respectively. siRNA knock-down of Fzd7 resulted in reduced DAB2 levels, and the overexpression activated a TCF-LEF reporter, but neither approach affected differentiation. Our focus turned to how canonical WNT6 signaling was attenuated to allow PrE cells to form parietal endoderm (PE). Dkk1, encoding a WNT antagonist, was examined and results showed that its expression increased in F9 cells treated with retinoic acid (RA) or overexpressing Wnt6. F9 cells overexpressing human DKK1 or treated with DKK1-conditioned medium and then treated with RA failed to differentiate, indicating that a negative feedback loop involving WNT6 and DKK1 attenuates canonical WNT-β-catenin signaling, thereby allowing PE cells to differentiate.
Collapse
Affiliation(s)
- Gregory Golenia
- a Department of Biology, Molecular Genetics Unit, Western University, London, ON N6A 5B7, Canada
| | - Mohamed I Gatie
- a Department of Biology, Molecular Genetics Unit, Western University, London, ON N6A 5B7, Canada
| | - Gregory M Kelly
- a Department of Biology, Molecular Genetics Unit, Western University, London, ON N6A 5B7, Canada.,b Child Health Research Institute, London, ON N6C 2V5, Canada.,c Ontario Institute for Regenerative Medicine, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
147
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
148
|
Thiele S, Rachner TD, Rauner M, Hofbauer LC. WNT5A and Its Receptors in the Bone-Cancer Dialogue. J Bone Miner Res 2016; 31:1488-96. [PMID: 27355180 DOI: 10.1002/jbmr.2899] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/08/2016] [Accepted: 06/24/2016] [Indexed: 12/12/2022]
Abstract
Wnt signaling is critical for tumorigenesis and skeletal remodeling. However, its contribution to the formation of metastatic bone lesions remains poorly defined. One major challenge of unraveling its role in cancer progression is the high complexity of Wnt signaling, which includes numerous ligands, receptors, and inhibitors, with intricate biological effects and specific signaling pathways depending on the cellular context. In this perspective, we summarize the role of the noncanonical Wnt ligand WNT5A in the development and metastatic process of osteotropic cancer entities. We focus on its tumor-suppressive function in breast cancer, tumor promoting effects in melanoma, and ambiguous role in prostate cancer, and discuss potential challenges and opportunities that may be associated with targeting Wnt signaling for cancer therapy and treatment of bone metastases. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stefanie Thiele
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| | - Tilman D Rachner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| | - Martina Rauner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Healthy Aging, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| |
Collapse
|
149
|
A Second WNT for Old Drugs: Drug Repositioning against WNT-Dependent Cancers. Cancers (Basel) 2016; 8:cancers8070066. [PMID: 27429001 PMCID: PMC4963808 DOI: 10.3390/cancers8070066] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 06/24/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant WNT signaling underlies cancerous transformation and growth in many tissues, such as the colon, breast, liver, and others. Downregulation of the WNT pathway is a desired mode of development of targeted therapies against these cancers. Despite the urgent need, no WNT signaling-directed drugs currently exist, and only very few candidates have reached early phase clinical trials. Among different strategies to develop WNT-targeting anti-cancer therapies, repositioning of existing drugs previously approved for other diseases is a promising approach. Nonsteroidal anti-inflammatory drugs like aspirin, the anti-leprotic clofazimine, and the anti-trypanosomal suramin are among examples of drugs having recently revealed WNT-targeting activities. In total, 16 human-use drug compounds have been found to be working through the WNT pathway and show promise for their prospective repositioning against various cancers. Advances, hurdles, and prospects of developing these molecules as potential drugs against WNT-dependent cancers, as well as approaches for discovering new ones for repositioning, are the foci of the current review.
Collapse
|
150
|
Mammary Development and Breast Cancer: A Wnt Perspective. Cancers (Basel) 2016; 8:cancers8070065. [PMID: 27420097 PMCID: PMC4963807 DOI: 10.3390/cancers8070065] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
The Wnt pathway has emerged as a key signaling cascade participating in mammary organogenesis and breast oncogenesis. In this review, we will summarize the current knowledge of how the pathway regulates stem cells and normal development of the mammary gland, and discuss how its various components contribute to breast carcinoma pathology.
Collapse
|